Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Basic Res Cardiol ; 116(1): 25, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33847835

RESUMEN

We have previously shown that overexpression of SKI, an endogenous TGF-ß1 repressor, deactivates the pro-fibrotic myofibroblast phenotype in the heart. We now show that SKI also functions independently of SMAD/TGF-ß signaling, by activating the Hippo tumor-suppressor pathway and inhibiting the Transcriptional co-Activator with PDZ-binding motif (TAZ or WWTR1). The mechanism(s) by which SKI targets TAZ to inhibit cardiac fibroblast activation and fibrogenesis remain undefined. A rat model of post-myocardial infarction was used to examine the expression of TAZ during acute fibrogenesis and chronic heart failure. Results were then corroborated with primary rat cardiac fibroblast cell culture performed both on plastic and on inert elastic substrates, along with the use of siRNA and adenoviral expression vectors for active forms of SKI, YAP, and TAZ. Gene expression was examined by qPCR and luciferase assays, while protein expression was examined by immunoblotting and fluorescence microscopy. Cell phenotype was further assessed by functional assays. Finally, to elucidate SKI's effects on Hippo signaling, the SKI and TAZ interactomes were captured in human cardiac fibroblasts using BioID2 and mass spectrometry. Potential interactors were investigated in vitro to reveal novel mechanisms of action for SKI. In vitro assays on elastic substrates revealed the ability of TAZ to overcome environmental stimuli and induce the activation of hypersynthetic cardiac myofibroblasts. Further cell-based assays demonstrated that SKI causes specific proteasomal degradation of TAZ, but not YAP, and shifts actin cytoskeleton dynamics to inhibit myofibroblast activation. These findings were supported by identifying the bi-phasic expression of TAZ in vivo during post-MI remodeling and fibrosis. BioID2-based interactomics in human cardiac fibroblasts suggest that SKI interacts with actin-modifying proteins and with LIM Domain-containing protein 1 (LIMD1), a negative regulator of Hippo signaling. Furthermore, we found that LATS2 interacts with TAZ, whereas LATS1 does not, and that LATS2 knockdown prevented TAZ downregulation with SKI overexpression. Our findings indicate that SKI's capacity to regulate cardiac fibroblast activation is mediated, in part, by Hippo signaling. We postulate that the interaction between SKI and TAZ in cardiac fibroblasts is arbitrated by LIMD1, an important intermediary in focal adhesion-associated signaling pathways. This study contributes to the understanding of the unique physiology of cardiac fibroblasts, and of the relationship between SKI expression and cell phenotype.


Asunto(s)
Fibroblastos/metabolismo , Insuficiencia Cardíaca/metabolismo , Vía de Señalización Hippo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Remodelación Ventricular , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/patología , Fibrosis , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/genética , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Fenotipo , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Sprague-Dawley , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/metabolismo
2.
Cell Tissue Res ; 385(3): 753-768, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34057573

RESUMEN

Fibroblast growth factor 2 (FGF2), produced as high (Hi-) and low (Lo-) molecular weight isoforms, is implicated in cardiac response to injury. The role of endogenous FGF2 isoforms during chronic stress is not well defined. We investigated the effects of endogenous Hi-FGF2 in a mouse model of simulated pressure-overload stress achieved by transverse aortic constriction (TAC) surgery. Hi-FGF2 knockout mice, expressing only Lo-FGF2, FGF2(Lo), and wild-type mice, FGF2(WT), expressing both Hi-FGF2 and Lo-FGF2, were used. By echocardiography, a decline in systolic function was observed in FGF2(WT) but not FGF2(Lo) mice compared to corresponding sham-operated animals at 4-8 weeks post-TAC surgery. TAC surgery increased markers of myocardial stress/damage including B-type natriuretic peptide (BNP) and the pro-cell death protein BCL2/adenovirus E1B 19 kDa protein-interacting protein-3 (Bnip3) in FGF2(WT) but not FGF2(Lo) mice. In FGF2(Lo) mice, cardiac levels of activated FGF receptor 1 (FGFR1), and downstream signals, including phosphorylated mTOR and p70S6 kinase, were elevated post-TAC. Finally, NR1D1 (nuclear receptor subfamily 1 group D member 1), implicated in cardioprotection from pressure-overload stress, was downregulated or upregulated in the presence or absence, respectively, of Hi-FGF2 expression, post-TAC surgery. In wild-type cardiomyocyte cultures, endothelin-1 (added to simulate pressure-overload signals) caused NR1D1 downregulation and BNP upregulation, similar to the effect of TAC surgery on the FGF2(WT) mice. The NR1D1 agonist SR9009 prevented BNP upregulation, simulating post-TAC findings in FGF2(Lo) mice. We propose that elimination of Hi-FGF2 is cardioprotective during pressure-overload by increasing FGFR1-associated signaling and NR1D1 expression.


Asunto(s)
Presión Sanguínea/genética , Factor 2 de Crecimiento de Fibroblastos/uso terapéutico , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Animales , Masculino , Ratones , Ratones Noqueados , Ratas , Transducción de Señal
3.
J Mol Cell Cardiol ; 126: 140-142, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30502349

RESUMEN

The functional significance of having two nuclei in a cell is unknown. Having two stores of genetic material may be advantageous for cell growth. Nuclear protein import is at a critical juncture in the cell to modify cell growth. This study addressed the potential for differential nuclear protein import in two nuclei of the same cell. Isolated adult rat cardiomyocytes were microinjected with an exogenous fluorescent protein conjugated with nuclear localization amino acid sequences to facilitate nuclear import and its detection. Our results demonstrate the rate of nuclear protein import was not significantly different between the two nuclei in the same cell. These data demonstrate that the two nuclei are functionally similar in a binucleated cardiomyocyte, at least as far as nucleocytoplasmic transport is concerned.


Asunto(s)
Núcleo Celular/metabolismo , Miocitos Cardíacos/metabolismo , Transporte Activo de Núcleo Celular , Animales , Masculino , Microinyecciones , Ratas Sprague-Dawley
4.
Am J Physiol Heart Circ Physiol ; 316(2): H279-H288, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30412444

RESUMEN

Cardiac fibroblast growth factor 2 (FGF2) exerts multiple paracrine activities related to cardiac response to injury. Endogenous FGF2 is composed of a mixture of 70% high- and 30% low-molecular-weight isoforms (Hi-FGF2 and Lo-FGF2, respectivley); although exogenously added Lo-FGF2 is cardioprotective, the roles of endogenous Hi-FGF2 or Lo-FGF2 have not been well defined. Therefore, we investigated the effect of elimination of Hi-FGF2 expression on susceptibility to acute cardiac damage in vivo caused by an injection of the genotoxic drug doxorubicin (Dox). Mice genetically depleted of endogenous Hi-FGF2 and expressing only Lo-FGF2 [FGF2(Lo) mice] were protected from the Dox-induced decline in ejection fraction displayed by their wild-type FGF2 [FGF2(WT)] mouse counterparts, regardless of sex, as assessed by echocardiography for up to 10 days post-Dox treatment. Because cardiac FGF2 is produced mainly by nonmyocytes, we next addressed potential contribution of fibroblast-produced FGF2 on myocyte vulnerability to Dox. In cocultures of neonatal rat cardiomyocytes (r-cardiomyocytes) with mouse fibroblasts from FGF2(WT) or FGF2(Lo) mice, only the FGF2(Lo)-fibroblast cocultures protected r-cardiomyocytes from Dox-induced mitochondrial and cellular damage. When r-cardiomyocytes were cocultured with or exposed to conditioned medium from human fibroblasts, neutralizing antibodies for human Hi-FGF-2, but not total FGF2, mitigated Dox-induced injury of cardiomyocytes. We conclude that endogenous Hi-FGF2 reduces cardioprotection by endogenous Lo-FGF2. Antibody-based neutralization of endogenous Hi-FGF2 may offer a prophylactic treatment against agents causing acute cardiac damage. NEW & NOTEWORTHY Cardiomyocytes, in vivo and in vitro, were protected from the deleterious effects of the anticancer drug doxorubicin by the genetic elimination or antibody-based neutralization of endogenous paracrine high-molecular-weight fibroblast growth factor 2 isoforms. These findings have a translational potential for mitigating doxorubicin-induced cardiac damage in patients with cancer by an antibody-based treatment.


Asunto(s)
Doxorrubicina/toxicidad , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Corazón/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miofibroblastos/metabolismo , Animales , Gasto Cardíaco , Cardiotoxicidad , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Corazón/fisiología , Humanos , Masculino , Ratones , Ratas
5.
Cell Tissue Res ; 374(3): 607-617, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30159756

RESUMEN

Doxorubicin (Dox)-induced cardiotoxicity, a limiting factor in the use of Dox to treat cancer, can be mitigated by the mitogenic factor FGF2 in vitro, via a heme oxygenase 1 (HO-1)-dependent pathway. HO-1 upregulation was reported to require protein kinase CK2 activity. We show that a mutant non-mitogenic FGF2 (S117A-FGF2), which does not activate CK2, is cardioprotective against acute cardiac ischemic injury. We now investigate the potential of S117A-FGF2 to protect cardiomyocytes against acute Dox injury and decrease Dox-induced upregulation of oxidized phospholipids. The roles of CK2 and HO-1 in cardiomyocyte protection are also addressed.Rat neonatal cardiomyocyte cultures were used as an established in vitro model of acute Dox toxicity. Pretreatment with S117A-FGF2 protected against Dox-induced: oxidative stress; upregulation of fragmented and non-fragmented oxidized phosphatidylcholine species, measured by LC/MS/MS; and cardiomyocyte injury and cell death measured by LDH release and a live-dead assay. CK2 inhibitors (TBB and Ellagic acid), did not affect protection by S117A-FGF2 but prevented protection by mitogenic FGF2. Furthermore, protection by S117A-FGF2, unlike that of FGF2, was not prevented by HO-1 inhibitors and S117A-FGF2 did not upregulate HO-1. Protection by S117A-FGF2 required the activity of FGF receptor 1 and ERK.We conclude that mitogenic and non-mitogenic FGF2 protect from acute Dox toxicity by common (FGFR1) and distinct, CK2/HO-1- dependent or CK2/HO-1-independent (respectively), pathways. Non-mitogenic FGF2 merits further consideration as a preventative treatment against Dox cardiotoxicity.


Asunto(s)
Cardiotónicos/farmacología , Quinasa de la Caseína II/metabolismo , Citoprotección/efectos de los fármacos , Doxorrubicina/toxicidad , Factor 2 de Crecimiento de Fibroblastos/farmacología , Hemo-Oxigenasa 1/metabolismo , Miocitos Cardíacos/patología , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Fosfolípidos/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
6.
J Mol Endocrinol ; 73(2)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38832641

RESUMEN

The objective was to assess the potential differential effects of human versus mouse growth hormone in vivo, given that human unlike mouse growth hormone can bind prolactin as well as the growth hormone receptor. To this end, a transgenic CD-1 mouse expressing human but not mouse growth hormone was generated, and the phenotypes of male mice fed with a regular chow or high-fat diet were assessed. Pancreas and epididymal white adipose tissue gene expression and/or related function were targeted as the pancreas responds to both prolactin and growth hormone receptor signaling, and catabolic effects like lipolytic activity are more directly attributable to growth hormone and growth hormone receptor signaling. The resulting human growth hormone-expressing mice are smaller than wild-type CD-1 mice, despite higher body fat and larger adipocytes, but both mouse types grow at the same rate with similar bone densities. Unlike wild-type mice, there was no significant delay in glucose clearance in human growth hormone-expressing mice when assessed at 8 versus 24 weeks on a high-fat diet. However, both mouse types showed signs of hepatic steatosis that correlated with elevated prolactin but not growth hormone RNA levels. The larger adipocytes in human growth hormone-expressing mice were associated with modified leptin (higher) and adiponectin (lower) RNA levels. Thus, while limited to observations in the male, the human growth hormone-expressing mice exhibit signs of growth hormone insufficiency and adipocyte dysfunction as well as an initial resistance to the negative effects of high-fat diet on glucose clearance.


Asunto(s)
Tejido Adiposo , Dieta Alta en Grasa , Hígado Graso , Glucosa , Homeostasis , Resistencia a la Insulina , Ratones Transgénicos , Animales , Humanos , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/genética , Hígado Graso/metabolismo , Hígado Graso/etiología , Hígado Graso/genética , Ratones , Masculino , Glucosa/metabolismo , Tejido Adiposo/metabolismo , Hormona de Crecimiento Humana/metabolismo , Hormona de Crecimiento Humana/genética , Hormona del Crecimiento/metabolismo , Hormona del Crecimiento/genética , Prolactina/metabolismo , Leptina/metabolismo , Adipocitos/metabolismo , Tejido Adiposo Blanco/metabolismo
7.
Biochim Biophys Acta ; 1818(8): 2009-13, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21763271

RESUMEN

The fundamental role played by connexins including connexin43 (Cx43) in forming intercellular communication channels (gap junctions), ensuring electrical and metabolic coupling between cells, has long been recognized and extensively investigated. There is also increasing recognition that Cx43, and other connexins, have additional roles, such as the ability to regulate cell proliferation, migration, and cytoprotection. Multiple phosphorylation sites, targets of different signaling pathways, are present at the regulatory, C-terminal domain of Cx43, and contribute to constitutive as well as transient phosphorylation Cx43 patterns, responding to ever-changing environmental stimuli and corresponding cellular needs. The present paper will focus on Cx43 in the heart, and provide an overview of the emerging recognition of a relationship between Cx43, its phosphorylation pattern, and development of resistance to injury. We will also review our recent work regarding the role of an enhanced phosphorylation state of Cx43 in cardioprotection. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.


Asunto(s)
Conexina 43/fisiología , Miocardio/metabolismo , Animales , Crioprotectores/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Corazón/fisiología , Cardiopatías/metabolismo , Humanos , Isquemia/metabolismo , Modelos Biológicos , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/citología , Fosforilación , Proteína Quinasa C/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Transducción de Señal
8.
Biochim Biophys Acta ; 1823(10): 1731-44, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22705883

RESUMEN

Connexin-43 (Cx43) is a membrane phosphoprotein that mediates direct inter-cellular communication by forming gap junctions. In this way Cx43 can influence gene expression, differentiation and growth. Its role in adipogenesis, however, is poorly understood. In this study, we established that Cx43 becomes highly phosphorylated in early adipocyte differentiation and translocates to the plasma membrane from the endoplasmic reticulum. As preadipocytes differentiate, Cx43 phosphorylation declines, the protein is displaced from the plasma membrane, and total cellular levels are reduced via proteosomal degradation. Notably, we show that inhibiting Cx43 degradation or constitutively over-expressing Cx43 blocks adipocyte differentiation. These data reveal that transient activation of Cx43 via phosphorylation followed by its degradation is vital for preadipocyte differentiation and maturation of functional adipocytes.


Asunto(s)
Adipogénesis , Conexina 43/metabolismo , Proteolisis , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Tejido Adiposo/citología , Animales , Citometría de Barrido por Láser , Leupeptinas/farmacología , Ratones , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo
9.
J Neuroendocrinol ; : e13258, 2023 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-36989439

RESUMEN

Maternal obesity is a serious health concern because it increases risks of neurological disorders, including anxiety and peripartum depression. In mice, a high fat diet (HFD) in pregnancy can negatively affect placental structure and function as well as maternal behavior reflected by impaired nest building and pup-retrieval. In humans, maternal obesity in pregnancy is associated with reduced placental lactogen (PL) gene expression, which has been linked to a higher risk of depression. PL acting predominantly through the prolactin receptor maintains energy homeostasis and is a marker of placenta villous trophoblast differentiation during pregnancy. Impaired neurogenesis and low serum levels of brain-derived neurotrophic factor (BDNF) have also been implicated in depression. Augmented neurogenesis in brain during pregnancy was reported in the subventricular zone (SVZ) of mice at gestation day 7 and linked to increased prolactin receptor signaling. Here, we used transgenic CD-1 mice that express human (h) PL during pregnancy to investigate whether the negative effects of diet on maternal behavior are mitigated in these (CD-1[hGH/PL]) mice. Specifically, we examined the effect of a HFD on nest building prepartum and pup retrieval postpartum, as well as on brain BDNF levels and neurogenesis. In contrast to wild-type CD-1[WT]mice, CD-1[hGH/PL] mice displayed significantly less anxiety-like behavior, and showed no impairment in prepartum nest building or postpartum pup-retrieval when fed a HFD. Furthermore, the HFD decreased prepartum and increased postpartum BDNF levels in CD-1[WT] but not CD-1[hGH/PL] mice. Finally, neurogenesis in the SVZ as well as phosphorylated mitogen-activated protein kinase, indicative of lactogenic signaling, appeared unaffected by pregnancy and diet at gestation day 7 in CD-1[hGH/PL] mice. These observations indicate that CD-1[hGH/PL] mice are resistant to the negative effects of HFD reported for CD-1[WT] mice, including effects on maternal behaviors and BDNF levels, and potentially, neurogenesis. This difference probably reflects a direct or indirect effect of the products of the hGH/PL transgene.

10.
Mol Cell Biochem ; 357(1-2): 1-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21556823

RESUMEN

Fibroblast growth factor-2 (FGF-2) is implicated in cardioprotection. However, previously we found that chronic elevation in cardiac FGF-2 levels in transgenic mice was associated with exaggerated, cyclosporine A-preventable, cellular infiltration after isoproterenol-induced injury, suggestive of an adverse outcome, although this was not examined with functional studies. We have now used highly sensitive tissue Doppler imaging (TDI) to evaluate cardiac functional parameters after isoproterenol administration in transgenic mice overexpressing the 18 kDa FGF-2 in the heart in vivo. Cardiac function was assessed in conscious FGF-2 transgenic and non-transgenic mice at 24 h as well as 2 and 4 weeks after isoproterenol administration, and in the absence or presence of either cyclosporine A or anti-CD3ε treatments. Isoproterenol decreased left ventricular endocardial velocity and strain rate by 47-51% at 24 h in non-transgenic mice, but to a significantly lesser extent (by 24%) in transgenic mice. While additional decreases were seen in non-transgenic mice at 2 weeks, there was no further reduction in ventricular endocardial velocity or strain rate up to 4 weeks post-treatment in FGF-2 transgenic mice. Functional improvement at 2 and 4 weeks post-isoproterenol was reduced significantly by treatment with cyclosporine A but not anti-CD3ε; the latter targets T lymphocyte activation more specifically. TDI values in the presence of chronic FGF-2 overexpression are prognostic of an improved cardiac outcome and protection from isoproterenol induced cardiac dysfunction in vivo. Our data also suggest that cyclosporine A-sensitive infiltrating cell population(s) may contribute to the sustained beneficial effect of FGF-2 in vivo.


Asunto(s)
Ciclosporina/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Disfunción Ventricular/inducido químicamente , Disfunción Ventricular/metabolismo , Agonistas Adrenérgicos beta/farmacología , Animales , Factor 2 de Crecimiento de Fibroblastos/genética , Isoproterenol/farmacología , Ratones , Ratones Transgénicos , Ultrasonografía Doppler , Disfunción Ventricular/diagnóstico por imagen
11.
Dev Dyn ; 239(6): 1573-84, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20503355

RESUMEN

In fibrosing hearts, myofibroblasts are associated with cardiac extracellular matrix remodeling. Expression of key genes in the transition of cardiac fibroblast to myofibroblast phenotype in post-myocardial infarction heart and in vitro has not been well addressed. Contractile, focal adhesion-associated, receptor proteins, fibroblast growth factor-2 (FGF-2) expression, and motility were compared to assess phenotype in adult and neonatal rat cardiac fibroblasts and myofibroblasts. Neonatal and adult fibroblasts undergo phenotypic transition to myofibroblastic cells, marked by increased alpha-smooth muscle actin (alphaSMA), smooth muscle myosin heavy chain (SMemb), extra domain-A (ED-A) fibronectin, paxillin, tensin, FGF-2, and TbetaRII receptor. Elevated ED-A fibronectin confirmed fibroblast to supermature myofibroblastic phenotype transition. Presence of myofibroblasts in vivo was noted in sections of healed infarct scar after myocardial infarction, and their expression is similar to that in culture. Thus, cultured neonatal and adult cardiac fibroblasts transition to myofibroblasts in vitro and share expression profiles of cardiac myofibroblasts in vivo. Reduced motility with in vitro passage reflects enhanced production of focal adhesions.


Asunto(s)
Fibroblastos/metabolismo , Adhesiones Focales/metabolismo , Músculo Liso/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular/fisiología , Movimiento Celular , Matriz Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/citología , Fibronectinas/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Ventrículos Cardíacos/metabolismo , Masculino , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Sprague-Dawley
12.
Cells ; 10(10)2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34685716

RESUMEN

Mitochondria, abundant organelles in high energy demand cells such as cardiomyocytes, can determine cell death or survival by regulating the opening of mitochondrial permeability transition pore, mPTP. We addressed the hypothesis that the growth factor FGF2, known to reside in intracellular locations, can directly influence mitochondrial susceptibility to mPTP opening. Rat cardiac subsarcolemmal (SSM) or interfibrillar (IFM) mitochondrial suspensions exposed directly to rat 18 kDa low molecular weight (Lo-) FGF2 isoform displayed increased resistance to calcium overload-induced mPTP, measured spectrophotometrically as "swelling", or as cytochrome c release from mitochondria. Inhibition of mitochondrial protein kinase C epsilon abrogated direct Lo-FGF2 mito-protection. Exposure to the rat 23 kDa high molecular weight (Hi) FGF2 isoform promoted cytochrome c release from SSM and IFM under nonstressed conditions. The effect of Hi-FGF2 was prevented by mPTP inhibitors, pre-exposure to Lo-FGF2, and okadaic acid, a serine/threonine phosphatase inhibitor. Western blotting and immunoelectron microscopy pointed to the presence of immunoreactive FGFR1 in cardiac mitochondria in situ. The direct mito-protective effect of Lo-FGF2, as well as the deleterious effect of Hi-FGF2, were prevented by FGFR1 inhibitors and FGFR1 neutralizing antibodies. We propose that intracellular FGF2 isoforms can modulate mPTP opening by interacting with mito-FGFR1 and relaying isoform-specific intramitochondrial signal transduction.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Mitocondrias Cardíacas/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Calcio/metabolismo , Citocromos c/metabolismo , Masculino , Mitocondrias Cardíacas/ultraestructura , Isoformas de Proteínas/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Ratas Sprague-Dawley , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores
13.
J Nutr ; 140(8): 1438-44, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20554900

RESUMEN

A high-lipid diet (HLD) may lead to adverse left ventricular (LV) remodeling and endothelial dysfunction in conditions of hemodynamic stress. Although congenital absence of nitric oxide synthase 3 (NOS3) leads to adverse LV remodeling after transverse aortic constriction (TAC), the effects of a HLD in this state remains unknown. Wild-type (WT) and NOS3 knockout mice (NOS3(-/-)) were randomized into the following 4 groups: 1) WT + low-lipid diet (LLD) (10% of energy); 2) WT + HLD (60% of energy); 3) NOS3(-/-) + LLD; and 4) NOS3(-/-) + HLD for a total of 12 wk. After 1 wk of randomization, TAC was performed on all groups. Serial echocardiography revealed a decrease in LV ejection fraction (LVEF) in WT and NOS3(-/-) mice fed the HLD compared with those fed the LLD diet at 12 wk post-TAC. Mice fed the NOS3(-/-) + HLD diet had a lower LVEF compared with mice in the other 3 groups (P < 0.05). There was greater myocyte hypertrophy, interstitial fibrosis, and percentage change in plasma cholesterol concentrations in the NOS3(-/-) + HLD group 12 wk post-TAC compared with the other 3 groups. Although high molecular weight fibroblast growth factor-2, a marker of cardiac hypertrophy, was more upregulated in the NOS3(-/-) + HLD group than in the other groups, markers of the renin-angiotensin system did not differ among them. A HLD potentiates LV dysfunction in NOS3(-/-) mice in a chronic pressure overload state.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/efectos adversos , Hipertensión/complicaciones , Óxido Nítrico Sintasa de Tipo III/deficiencia , Disfunción Ventricular Izquierda/etiología , Animales , Aorta , Presión Sanguínea , Colesterol/sangre , Constricción , Ecocardiografía , Ingestión de Energía , Factor 2 de Crecimiento de Fibroblastos/análisis , Ventrículos Cardíacos/patología , Hipertrofia Ventricular Izquierda/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peso Molecular , Células Musculares/patología , Miocardio/patología , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/fisiología , Volumen Sistólico , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/patología
14.
FEBS J ; 287(5): 1005-1034, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31545550

RESUMEN

Temozolomide (TMZ) is a chemotherapy agent used to treat Grade IV astrocytoma, also known as glioblastoma (GBM). TMZ treatment causes DNA damage that results in tumor cell apoptosis and increases the survival rate of GBM patients. However, chemoresistance as a result of TMZ-induced autophagy significantly reduces this anticancer effects over time. Statins are competitive inhibitors of HMG-CoA reductase, the rate-limiting enzyme of the mevalonate (MEV) cascade. Statins are best known for their cholesterol (CH)-lowering effect. Long-term consumption of statins, prior to and in parallel with other cancer therapeutic approaches, has been reported to increase the survival rate of patients with various forms of cancers. In this study, we investigated the potentiation of TMZ-induced apoptosis by simvastatin (Simva) in human GBM cell lines and patient GBM cells, using cell monolayers and three-dimensional cell culture systems. The incubation of cells with a combination of Simva and TMZ resulted in a significant increase in apoptotic cells compared to cells treated with TMZ alone. Incubation of cells with CH or MEV cascade intermediates failed to compensate the decrease in cell viability induced by the combined Simva and TMZ treatment. Simva treatment inhibited the autophagy flux induced by TMZ by blocking autophago-lysosome formation. Our results suggest that Simva sensitizes GBM cells to TMZ-induced cell death in a MEV cascade-independent manner and identifies the inhibition of autophagosome-lysosome fusion as a promising therapeutic strategy in the treatment of GBM.


Asunto(s)
Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Muerte Celular/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Simvastatina/farmacología , Temozolomida/farmacología , Animales , Línea Celular Tumoral , Femenino , Glioblastoma/metabolismo , Humanos , Macrólidos/farmacología , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Can J Physiol Pharmacol ; 87(10): 798-804, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19898562

RESUMEN

Fibroblast growth factor 2 (FGF-2) is a multifunctional protein translated as high and low molecular weight isoforms (hi- and lo-FGF-2, respectively). Although the postconditioning cardioprotective effect of lo-FGF-2 (18 kDa) has been documented, hi-FGF-2 is less well studied. We used an isolated perfused rat heart model of ischemia-reperfusion to study the effects of postischemic (during reperfusion) administration of hi-FGF-2 on recovery of contractile function and tissue salvage, as indicated by decreased cytosolic cytochrome c levels. Compared with the vehicle-treated group, hi-FGF-2-treated hearts had significantly improved recovery of systolic pressure, developed pressure, rates of contraction and relaxation, and coronary flow, as well as decreased relative levels of cytosolic cytochrome c. The effects of hi-FGF-2 on functional recovery and cytosolic cytochrome c were indistinguishable from those induced by lo-FGF-2. Both hi- and lo-FGF-2 upregulated relative levels of phosphorylated (activated) Akt and p70 S6 kinase, and they both promoted translocation of alpha, epsilon, and zeta isoforms of protein kinase C (PKC) to the particulate fraction of reperfused hearts. The magnitude of the effect on PKCzeta and p70 S6 kinases, however, was significantly more potent in the hi-FGF-2 than in the lo-FGF-2 group. We conclude that acute postischemic cardioprotection by hi- or lo-FGF-2 is isoform nonspecific and likely to be mediated by PKC and Akt. Nevertheless, isoform-specific functions are suggested by the augmented sensitivity of p70 S6 and PKCzeta to hi-FGF-2.


Asunto(s)
Cardiotónicos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Cardiopatías/prevención & control , Precondicionamiento Isquémico Miocárdico , Proteína Oncogénica v-akt/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Animales , Western Blotting , Activación Enzimática/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/química , Técnicas In Vitro , Isoenzimas/metabolismo , Masculino , Peso Molecular , Daño por Reperfusión Miocárdica/patología , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo
16.
Front Mol Neurosci ; 12: 328, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32038161

RESUMEN

Astrocytes are the most abundant type of glial cells in the brain, and they play a key role in Alzheimer's disease (AD). Fibroblast Growth Factor-2 (FGF-2) has been implicated as a potential therapeutic agent for treating AD. In the present study, we investigated the protective effects of low molecular weight (LMW; 17 KDa) and high molecular weight (HMW; 23 KDa) forms of FGF-2 on Aß1-42-induced toxicity, and proliferation in astrocytes. We show that both isoforms of FGF-2 have similar protective effects against Aß1-42-induced cytotoxicity in primary cultured cortical astrocytes as measured by Lactate Dehydrogenase (LDH) release assay. Additionally, 17 KDa FGF-2 significantly promoted astrocyte proliferation as measured by Trypan Blue, DRAQ5 and 5-ethynyl-2'-deoxyuridine (EdU) staining, but not 23 kDa FGF-2. Furthermore, our results demonstrated that AKT signaling pathway was required for the protective and proliferative effects of FGF-2. Downstream effector studies indicated that 17 kDa FGF-2 promoted astrocyte proliferation by enhanced expression of c-Myc, Cyclin D1, Cyclin E. Furthermore, our data suggested that Cyclin D1 was required for the proliferative effect of LMW FGF2 in astrocytes. Taken together, our findings provide important information for the similarities and differences between 23 kDa and17 kDa isoforms of FGF-2 on astrocyte survival and proliferation.

17.
Chem Biol Interact ; 303: 35-39, 2019 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-30707978

RESUMEN

Doxorubicin (Dox), a widely used chemotherapy drug, can also cause cardiotoxic effects leading to heart failure. The excessive oxidative stress caused by Dox results in the modification of a variety of cellular molecules, including phospholipids. In cardiomyocytes, Dox increases oxidation of a species of phospholipids, phosphatidylcholine, which has been associated with increased cell death. Oxidized phospholipids (Ox-PL) are involved in development and progression of various pathologies, including atherosclerosis, thrombosis, and tissue inflammation. Moreover, Ox-PL and excess iron are associated with ferroptosis, a form of regulated cell death. Neutralizing Ox-PL increases resistance to ischemia-reperfusion injuries which is linked to preservation of the mitochondrial membrane potential. This review aims to discuss the potential role of Ox-PL in Dox-induced pathology and supports the notion that a better understanding of the field could point to new strategies to prevent cardiotoxicity.


Asunto(s)
Cardiotoxicidad/etiología , Doxorrubicina/efectos adversos , Fosfolípidos/metabolismo , Animales , Humanos , Oxidación-Reducción , Fosfolípidos/fisiología
18.
Prog Biophys Mol Biol ; 94(1-2): 245-64, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17462721

RESUMEN

The purpose of this paper is to provide a brief overview of current thinking on the role of connexins, in particular Cx43, in growth regulation, and a more detailed discussion as to potential mechanisms involved with an emphasis on gene expression. While the precise molecular mechanism by which connexins can affect the growth of normal or tumor cells remains elusive, a number of exciting reports have expanded our understanding and are presented in some detail. Thus, we will discuss (Section 2): the role of protein-protein interactions in integrating connexins into multiple signal transduction pathways; phosphorylation at specific sites and reversal of growth inhibition; the role of the carboxy-terminal regulatory domain as a signaling molecule. Some of our latest work on the potential functions of endogenously produced carboxy-terminal fragments of Cx43 are also presented (Section 3). Finally, Section 4 will pay tribute to the rapidly emerging realization that connexins such as Cx43 and Cx32 exert important and extensive effects on gene expression, particularly those genes linked to growth regulation.


Asunto(s)
Adaptación Fisiológica/fisiología , Proliferación Celular , Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Regulación de la Expresión Génica/fisiología , Transducción de Señal/fisiología , Animales , Humanos , Modelos Biológicos
19.
DNA Cell Biol ; 37(11): 866-877, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30230915

RESUMEN

The fibroblast growth factor (FGF) 16 gene is preferentially expressed by cardiomyocytes after birth with levels increasing into adulthood. Null mice and isolated heart studies suggest a role for FGF-16 in cardiac maintenance and survival, including increased resistance to doxorubicin (DOX)-induced injury. A single treatment with DOX was also shown to rapidly deplete endogenous rat FGF-16 mRNA at 6 h in both adult heart and neonatal cardiomyocytes. However, the effect of DOX on rat cardiac function at the time of decreased FGF-16 gene expression and the effect of FGF-16 availability on cardiomyocyte survival, including in the context of acute DOX cytotoxicity, have not been reported. The objective was to assess the effect of acute (6 and 24 h) DOX treatment on cardiac function and the effects of FGF-16 small interfering RNA "knockdown," as well as adenoviral overexpression, in the context of acute DOX cytotoxicity, including cardiomyocyte survival and DOX efflux transport. A significant decrease in heart systolic function was detected by echocardiography in adult rats treated with 15 mg DOX/kg at 6 h; however, unlike FGF-16, there was no change in atrial natriuretic peptide transcript levels. Both systolic and diastolic dysfunctions were observed at 24 h. In addition, specific FGF-16 "knockdown" in neonatal rat cardiomyocytes results in a significant increase in cell death. Conversely, adenoviral FGF-16 overexpression was associated with a significant decrease in cardiomyocyte injury as a result of 1 µM DOX treatment. A specific increase in efflux transporter gene expression and DOX efflux was also seen, which is consistent with a reduction in DOX cytotoxicity. Finally, the increased efflux and decreased DOX-induced damage with FGF-16 overexpression were blunted by inhibition of FGF receptor signaling. These observations are consistent with FGF-16 serving as an endogenous cardiomyocyte survival factor, which may involve a positive effect on regulating efflux transport to reduce cardiotoxicity.


Asunto(s)
Cardiomiopatías/genética , Citotoxinas/toxicidad , Doxorrubicina/toxicidad , Factores de Crecimiento de Fibroblastos/genética , Miocitos Cardíacos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Transporte Biológico/efectos de los fármacos , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/patología , Cardiomiopatías/prevención & control , Supervivencia Celular/efectos de los fármacos , Ecocardiografía , Factores de Crecimiento de Fibroblastos/agonistas , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Pruebas de Función Cardíaca , Inyecciones Intraperitoneales , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Cultivo Primario de Células , Pirroles/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
20.
Front Mol Neurosci ; 11: 377, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30369869

RESUMEN

The occurrence of depressive disorder has long been attributed to changes in monoamines, with the focus of drug treatment strategies being to change the effectiveness of monoamines. However, the success achieved by changing these processes is limited and further stimulates the exploration of alternative mechanisms and treatments. Fibroblast growth factor 2 (FGF-2), which occurs in a high-molecular weight (HMW) and low-molecular weight (LMW) form, is a potent developmental modulator and nervous system regulator that has been suggested to play an important role in various psychiatric disorders. In this study, we investigated the antidepressant effects of HMW and LMW FGF-2 on depression induced by chronic stress. Both peripheral LMW and HMW FGF-2 attenuated the depression-like behaviors in chronic unpredictable mild stress (CUMS) mice to a similar extent, as determined by the forced swimming, tail suspension, and sucrose preference tests. We then showed that CUMS-induced oxidative stresses in mice were inhibited by FGF-2 treatments both in central and peripheral. We also showed that both forms of FGF-2 increased the phosphorylation of ERK and AKT, increased Bcl-2 expression and inhibited caspase-3 activation in CUMS mice. Interestingly, HMW FGF-2 enhanced the activity of the brain-derived neurotrophic factor (BDNF) to a greater extent than did LMW FGF-2 in the hippocampus. Taken together, these results suggest that depressive symptoms can be relieved by administering different forms of FGF-2 peripherally in a CUMS-induced depression model through a similar antidepressant signaling pathway, therefore suggesting a potential clinical use for FGF-2 as a treatment for depression.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA